• Keine Ergebnisse gefunden

Development of functional assays for human neuropeptide Y (Y

N/A
N/A
Protected

Academic year: 2022

Aktie "Development of functional assays for human neuropeptide Y (Y"

Copied!
129
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

Development of functional assays for human neuropeptide Y (Y 1,2,4,5 ) receptors exploiting

GTPase activity and (bio)luminescence as readout

Dissertation

zur Erlangung des Doktorgrades der Naturwissenschaften (Dr. rer. nat.) der Naturwissenschaftlichen Fakultät IV – Chemie und Pharmazie –

der Universität Regensburg

vorgelegt von Nathalie Pop aus Sathmar

2010

(2)

Januar 2006 bis Juli 2010 unter der Leitung von Herrn Prof. Dr. Armin Buschauer, Herrn Prof. Dr. Günther Bernhardt und Herrn Prof. Dr. Roland Seifert am Institut für Pharmazie der Naturwissenschaftlichen Fakultät IV – Chemie und Pharmazie der Universität Regensburg.

Das Promotionsgesuch wurde eingereicht im Juli 2010.

Tag der mündlichen Prüfung: 06. August 2010

Prüfungsausschuss: Prof. Dr. J. Heilmann (Vorsitzender)

Prof. Dr. A. Buschauer (Erstgutachter) Prof. Dr. G. Bernhardt (Zweitgutachter) Prof. Dr. A. Göpferich (Drittprüfer)

(3)

Für meine Familie

„Wir wollen die Gegenwart mit Hoffnung gestalten, weil uns die Zukunft gewiss ist“

(Peter Hahne)

(4)

Herrn Prof. Dr. Armin Buschauer für die Möglichkeit an diesem spannenden und vielseitigen Projekt in seinem Arbeitskreis arbeiten zu dürfen, für seine Hilfsbereitschaft sowie seine konstruktive Kritik bei der Durchsicht dieser Arbeit.

Herrn Prof. Dr. Günther Bernhardt für seine fachliche Anleitung, sein Interesse an den Projekten, seine Hilfsbereitschaft und die kritische Durchsicht der Arbeit.

Herrn Prof. Dr. Roland Seifert (Medizinische Hochschule Hannover) für seine wissenschaftlichen Anregungen, sein Interesse sowie seine konstruktive Kritik bei der Entwicklung der GTPase assays für den Y2 und Y4 Rezeptor.

Frau Prof. Dr. Chiara Cabrele (Ruhruniversität Bochum) für die Synthese der benötigten Peptide, ihre Hilfsbereitschaft und die stets gute Zusammenarbeit.

Herrn Dr. Viacheslav O. Nikolaev (Universität Würzburg) für die Bereitstellung des Plasmids pcDNA3-EYFP-Epac2B(murine)-ECFP.

Herrn Dr. J. Daniels (Glaxo Wellcome) für die Bereitstellung des Liganden GW1229.

Herrn Dr. Dietmar Gross für die Einarbeitung in die Konfokalmikroskopie und seine stete Hilfsbereitschaft.

Frau Dr. Katharina Wenzel-Seifert für ihre Hilfsbereitschaft bei Fragen zur Klonierung und PCR.

Herrn Dr. Erich Schneider, Herrn Dr. David Schnell und Herrn Dr. Patrick Igel für ihre wertvolle Unterstützung und die anregenden Diskusionen vor allem auf dem Gebiet der GTPase.

Herrn Dr. Max Keller für die Aufreinigung von Peptiden mittels HPLC und die stets gute Zusammenarbeit.

Herrn Dr. Albert Brennauer und Herrn Nikola Pluym für die Bereitstellung von Y2

Antagonisten sowie Herrn Dr. Liantao Li für die Synthese des Y5 Antagonisten CGP 71683A.

Frau Dr. Anja Kraus und Herrn Dr. Patrick Igel für die Bereitstellung von Substanzen zur Testung am Y4 Rezeptor.

Frau Dr. Edith Hofinger und Herrn Dr. Ralf Ziemek für die freundliche Unterstützung bei molekularbiologischen Arbeiten und dem Erlernen der Zellkultur in der Anfangszeit der Promotion.

Frau Evi Schreiber, Frau Susanne Bollwein, Frau Brigitte Wenzel für die Unterstützung auf dem Gebiet der Zellkultur und die gute Zusammenarbeit.

Frau Gertraud Wilberg für die Unterstützung auf dem Gebiet der Sf9 Zellkultur, die Einarbeitung in die SDS-Page und Western Blot und für ihre Hilfsbereitschaft.

(5)

Frau Martina Wechler, Frau Silvia Heinrich, Frau Karin Reindl und Herrn Peter Richthammer für ihre Unterstützung bei organisatorischen und technischen Problemen.

meinen ForschungspraktikantInnen Verena Venjakob, Daniela Schlosser, Nina Kraupner und Stefan Weidinger für die Testung von Substanzen, Durchführung von Western Blots und PCRs sowie meinen Wahlpflichtstudentinnen Alexa Schenke und Carolin Lichtinger für die Durchführung von Kristallviolett assays, Stefanie Surner für die Durchführung von PCRs sowie Jessica Wagner und Barbara Fink für die Hilfe bei Membranpräparationen und Western Blots.

Frau Daniela Erdmann, Frau Janina Hamberger, Frau Irena Brunskole, Herrn Miroslaw Lopuch, Herrn Dr. Johannes Mosandl, Herrn Dr. Martin Göttle, Herrn Dr. Patrick Igel für ihre tatkräftige Unterstützung bei Membranpräparationen.

allen hier namentlich nicht genannten Kollegen unseres Lehrstuhls für ihre Kollegialität und Hilfsbereitschaft sowie auch der Nachbarlehrstühle für das gute Arbeitsklima.

dem Graduiertenkolleg 760 der DFG für die finanzielle Unterstützung und wissenschaftliche Förderung.

Desweiteren geht mein besonderer Dank an:

Daniela Erdmann, Janina Hamberger, Johannes Mosandl und Tobias Scheuerer für ihre Freundschaft und Geduld in den letzten gemeinsamen Jahren in Regensburg.

Peter Jarzyna, Martin Göttle, Dietmar Gross und Erich Schneider für die besondere Stütze, die sie mir während der Promotion waren.

Annette Holler und Karin Schmidbauer für ihre Freundschaft und ihr beständiges unglaubliches Interesse (als nicht-Naturwissenschaftlerinnen) an der Entwicklung meiner Arbeit.

Christina Wirth, Kristin Kölbl, Marlen Schmidt, Susanne Eichhorn und Johannes Wessel für die schöne gemeinsame „Wohnzeit“ in Regensburg.

meinen wunderbaren Eltern und meinem ebenso wunderbaren Bruder, Christian, für ihre beständige und selbstlose Unterstützung nicht nur in Zeiten der Promotion.

(6)
(7)

Publications

Keller, M., Pop, N., Hutzler, C., Beck-Sickinger, A.G., Bernhardt, G., Buschauer, A., Guanidine -acylguanidine bioisosteric approach in the design of radioligands: Synthesis of a tritium-labeled NG-propionylargininamide ([3H]-UR-MK114) as a highly potent and selective neuropeptide Y Y1 receptor antagonist. J. Med. Chem. (2008), doi: 10.1021/jm801018u

Poster presentations

Pop N., Seifert R., Bernhardt G., Buschauer A., Functional reconstitution of the human neuropeptide Y Y2 and Y4 receptors with Gi/o-proteins in Sf9 insect cells, Conference of the German Pharmaceutical Society (DPhG), Jena (Germany), September 2009

Pop N., Seifert R., Bernhardt G., Buschauer A., Functional reconstitution of the human neuropeptide Y Y4 receptor with Gi/o-proteins in Sf9 insect cells, 4th International Summer School “Medicinal Chemistry”, Regensburg (Germany), September 2008

Pop N., Bernhardt G., Buschauer A., Development of a FRET based cAMP assay for the human NPY Y1 receptor, Annual Meeting of the GDCh, Fachgruppe Medizinische Chemie, “Frontiers in Medicinal Chemistry”, Regensburg (Germany), March 2008 Keller M., Pop N., Schneider E., Hoefelschweiger B.K., Brennauer A., Gross D., Wolfbeis O.S., Bernhardt G., Dove S. and Buschauer A., Fluorescence labeled NPY Y1 receptor antagonists, Conference of the German Pharmaceutical Society (DPhG), Erlangen (Germany), October 2007, and Annual Meeting of the GDCh, Fachgruppe Medizinische Chemie, “Frontiers in Medicinal Chemistry”, Regensburg (Germany), March 2008 Lopuch M., Pop N., Keller M., Cwik M., Bernhardt G., Buschauer A., Stable expression of neuropeptide Y receptor type 1 tagged with cyan and yellow fluorescent protein – investigations using radiochemistry and fluorescence-based methods, 4th International Summer School “Medicinal Chemistry”, Regensburg (Germany), September 2008

(8)
(9)

Contents IX

Contents

1 General introduction ... 1

1.1 G protein-coupled receptors ... 1

1.2 The neuropeptide Y (NPY) multiligand/multireceptor system ... 2

1.2.1 The neuropeptide Y family of peptides ... 2

1.2.2 NPY receptors ... 3

1.2.3 NPY receptor ligands ... 5

1.3 Development of novel functional assays for NPY receptors ... 8

1.3.1 FRET based cAMP assay for the human NPY Y1 receptor ... 10

1.3.2 Fluorescence and luminescence based calcium assays for the human NPY Y5 receptor ... 11

1.3.3 Steady-state GTPase assay for the human NPY Y2 and Y4 receptors ... 12

1.4 References ... 13

2 Scope and Objectives ... 21

2.1 References ... 23

3  Towards a FRET based functional cAMP assay for the human NPY Y1 receptor in SK-N-MC cells ... 25 

3.1  Introduction ... 25 

3.2  Materials and Methods ... 27 

3.2.1  Materials ... 27 

3.2.2  Cell culture ... 27 

3.2.3  Chemosensitivity assay ... 27 

3.2.4  Transfection of SK-N-MC cells ... 28 

3.2.5  Spectrofluorimetry ... 29 

3.2.6  Cell sorting ... 29 

3.2.7  Confocal microscopy ... 30 

3.3  Results and discussion ... 30 

3.3.1  Spectrofluorimetry ... 30

3.3.2  Confocal microscopy ... 32 

(10)

3.4  Summary and conclusions ... 35 

3.5  References ... 36

4  Towards fluorescence and luminescence based functional assays for the human NPY Y5 receptor in HEC-1B-hY5 cells ... 37 

4.1  Introduction ... 37 

4.1.1  Redirecting receptor signalling via chimeric G proteins ... 37 

4.2  Materials and Methods ... 38 

4.2.1  Preparation of media and agar plates ... 38 

4.2.2  Preparation of competent E. coli ... 39 

4.2.3  Transformation of E. coli ... 39 

4.2.4  Preparation of plasmid DNA... 39 

4.2.4.1 Miniprep ... 39

4.2.4.2 Maxiprep ... 40

4.2.4.3 Determination of DNA concentration and sequencing ... 40

4.2.4.4 Restriction enzyme digestion and dephosphorylation of plasmid ends ... 41

4.2.4.5 Agarose gel electrophoresis ... 41

4.2.4.6 Purification of PCR product and recovery of DNA fragments from agarose gels ... 42

4.2.5  Preparation of chimeric Gαqi9 by PCR ... 42 

4.2.6  Subcloning of pcDNA3.1/Hygro-Gαqi9 ... 43 

4.2.7  Cell culture and chemosensitivity assay ... 44 

4.2.8  Transfection of HEC-1B-hY5 cells with pcDNA3.1/Hygro-Gαqi5/9 ... 45 

4.2.9  Transfection of HEC-1B-hY5-Gαqi5/9 with pcDNA3.1/Zeo-mtAEQ ... 46 

4.2.10  Flow cytometric calcium assay ... 46 

4.2.11  Spectrofluorimetric calcium assay ... 47 

4.2.12  Aequorin assay ... 48 

4.3  Results and discussion ... 49 

4.3.1  Flow cytometric calcium assay ... 49 

4.3.2  Spectrofluorimetric calcium assay ... 51 

4.3.3  Aequorin assay ... 52 

4.4  Summary and conclusions ... 53 

4.5  References ... 53

(11)

Contents XI

5  Establishment of a steady-state GTPase assay for the human NPY Y2 receptor ... 57 

5.1  Introduction ... 57 

5.1.1  Functional assays for the NPY Y2 receptor ... 57 

5.1.2  Coupling of the NPY Y2 receptor to Gi/o proteins ... 58 

5.2  Materials and Methods ... 59 

5.2.1  Preparation of the SF-hY2-His6 construct by sequential overlap extension PCR ... 59 

5.2.1.1  PCR 1a for the hY2R ... 61 

5.2.1.2  PCR 1b for the hY2R ... 62 

5.2.1.3  PCR 2 for the hY2R ... 63 

5.2.2  Subcloning of SF-hY2-His6 into pGEM-3Z ... 64 

5.2.3  Subcloning of SF-hY2-His6 into pVL1392 ... 65 

5.2.4  Sf9 cell culture ... 66 

5.2.5  Generation of recombinant baculoviruses ... 66 

5.2.6  Membrane preparation and determination of protein concentration ... 67 

5.2.7  SDS Page and immunoblot analysis ... 68 

5.2.8  Y2 receptor antagonists ... 71 

5.2.9  Steady-state GTPase assay ... 71 

5.3  Results and Discussion ... 73 

5.3.1  Immunoblot analysis of membranes ... 73 

5.3.2  Coupling efficiency of the hY2R to Gαi/o proteins ... 78 

5.3.3  Effects of monovalent salts on the GTPase activity ... 79 

5.3.4  Effect of N-glycosylation on receptor function ... 81 

5.3.5  Evaluation of the expression system hY2R + Gαi2 + Gβ1γ2 ... 82 

5.4  Summary and conclusions ... 83

5.5 References ... 84

6 Establishment of a steady-state GTPase assay for the human NPY Y4 receptor ... 87

6.1 Introduction ... 87

6.2 Materials and Methods ... 88

6.2.1 Preparation of SF-hY4R-His6 construct by overlap extension PCR ... 88

6.2.2 Y4 receptor ligands ... 90

6.3 Results and Discussion ... 91

(12)

6.3.1 Immunoblot analysis of membranes ... 91

6.3.1.1 Protein expression in membrane batch ... 91

6.3.1.2 Quantitative immunoblot analysis ... 93

6.3.1.3 Effect of N-glycosylation on the structure and the expression level of the hY4R ... 95

6.3.2 Coupling efficiency of the hY4R to Gαi/o proteins ... 97

6.3.3 Effects of monovalent salts on the GTPase activity ... 99

6.3.4 Effect of N-glycosylation on receptor function ... 101

6.3.5 Evaluation of the hY4R Sf9 expression systems ... 101

6.3.6 Screening of potential NPY Y4R ligands ... 105

6.4 Summary and conclusions ... 109

6.5 References ... 110

7  Summary ... 113 

(13)

Abbreviations XIII Abbreviations

aa amino acid

AC adenylyl cyclase

AcNPV Autographa californica nuclear polyhedrosis virus AKT family of protein kinases B

ATP adenosine 5`-triphosphate

bp base pair(s)

BRET bioluminescence resonance energy transfer

BSA bovine serum albumine

cAMP cyclic 3`:5`-adenosine monophosphate

cDNA copy DNA

CHO cells chinese hamster ovary cells CIP calf intestinal phosphatise

CNS central nervous system

DAG 1,2-diacylglycerol

DMSO dimethylsufloxide

DTT dithiothreitol

EC50 agonist concentration which induces 50 % of the maximum effect

EDTA ethylenediaminetetraacetic acid (Ca2+-chelator) Epac exchange protein directly activated by cAMP Emax efficacy (maximal response)

FACS fluorescence activated cell sorter

FCS fetal calf serum

Fl-1, FL-2, Fl-3, Fl-4 fluorescence channels of the flow cytometer FLAG octapeptide epitope for the labelling of proteins FLIPR fluorescence imaging plate reader FRET fluorescence resonance energy transfer

i1, Gαi2, Gαi3, Gαo1 α-subunits of the G-proteins that inhibit adenylyl cyclase Gαq/11 α-subunits of the G-proteins that stimulate phospholipase C Gαqi5 chimeric α-subunit of Gαq/11 and the carboxyl terminus of Gαi to

redirect the signalling of Gαi coupled receptors towards calcium release

Gβγ βγ-subunits of a heterotrimeric G-protein

GDP guanosine 5`-diphosphate

GPCR G protein-coupled receptor

GTP guanosine 5`-triphosphate

GTPγS guanosine 5`-[γ-thio]triphosphate HEC-1B cells human endometrial carcinoma cells HEK 293 cells human embryonic kidney cells

His6 hexahistidine tag

IC50 antagonist concentration which suppresses 50 % of an agonist induced effect

(14)

i.c.v. intracerebroventricular

i.p. intraperitoneal

IP3 inositoltriphosphate

i.v. intravenous LAF laminar air flow (for sterile working banks) LB lysogeny broth (for cultivation of E. coli) MAPK mitogen-activated protein kinase mtAEQ mitochondrially targeted aequorin

NPY neuropeptide Y

PBS phosphate buffered saline

PCR polymerase chain reaction

Pi inorganic phosphate

PLC phospholipase C

PMSF phenylmethylsulfonylfluoride

PP pancreatic polypeptide

PYY peptide YY

RGS regulator of G protein signalling

RIA radio immuno assay

s.c. subcoutaneous

SDS sodiumdodecylsulfate

SDS Page sodiumdodecylsulfate polyacrylamide gel electrophoresis S.E.M. standard error of the mean

Sf9 insect cell line of Spodoptera frugiperda

SOC salt optimized + carbon broth (for transformation of E. coli) SPA scintillation proximity assay

SRC proto-oncogenic tyrosin kinases

TAE tris-acetat-EDTA buffer

TBE tris-borat-EDTA buffer

TM transmembrane domain

Tris tris(hydroxymethyl)aminomehtan

Y1, Y2, Y3, Y4, Y5, y6, Y7 neuropeptide Y receptor subtypes

(15)

Chapter 1

1 General introduction

1.1 G protein-coupled receptors

G protein-coupled receptors (GPCRs) constitute the largest known gene superfamily (≈ 2 %) of the human genome with up to approximately 800 members. Over half of these representatives are olfactory, leaving ≈ 380 unique functional nonolfactory/nonsensory GPCRs of which about only 40 are known as targets of modern drugs. Nevertheless, up to 30 % of all marketed prescription drugs act on GPCRs (Jacoby et al., 2006; Lagerström and Schiöth, 2008). Thus, G protein-coupled receptors offer an interesting field with high potential for drug discovery.

Though, the physiological agonists are extremely versatile, ranging from light, ions, amines, peptides to proteins (Bockaert and Pin, 1999), GPCRs can be phylogenetically divided into five main families termed glutamate, rhodopsin, adhesion, frizzled/taste2 and secretin (Fredriksson et al., 2003). The common structural features of the G protein-coupled receptor superfamily are seven membrane-spanning helices connected by three alternating intracellular and extracellular loops and flanked by an extracellular N-terminus and an intracellular C- terminus, respectively. Due to the high abundance in retina the bovine rhodopsin was the first mammalian GPCR to be crystallized by Palczewski in 2000, providing the first insight into the three-dimensional architecture of a G protein-coupled receptor (Palczewski et al., 2000).

Further crystal structures have been resolved recently for the human β2-adrenoceptor (hβ2AR) (Rasmussen et al., 2007; Rosenbaum et al., 2007), the turkey β1AR (Warne et al., 2008) and the human adenosine 2A receptor (Jaakola et al., 2008), which was only possible by diverse receptor modifications like truncation, use of an antibody or construction of a receptor/T4- lysozyme fusion protein as stabilizing elements in combination with receptor bound inverse

(16)

agonists or antagonists. These efforts were amongst others performed to elucidate receptor conformation upon activation by ligand binding and coupling to heterotrimeric G proteins, the mediators of intracellular effects (cf. section 1.3). However, in contrast to their original denomination, GPCRs do not only signal via heterotrimeric G proteins, but interaction with β- arrestin after receptor phosphorylation is part of the major signaling process (Violin and Lefkowitz, 2007; Rajagopal et al., 2010), which comprises e.g. regulation of kinases - like MAPKs, SRC or AKT – and of the receptor itself by desensitization and internalization.

Therefore, GPCRs lately are also named seven transmembrane receptors (7TMRs). The present work focuses on GPCR signaling via heterotrimeric G proteins. A detailed illustration of the G protein activation/deactivation cycle is shown in section 1.3.

1.2 The neuropeptide Y (NPY) multiligand/multireceptor system

1.2.1 The neuropeptide Y family of peptides

Neuropeptide Y (NPY) represents together with peptide YY (PYY) and the pancreatic polypeptide (PP) a ligand family of neuroendocrine hormones, consisting of 36 amino acids each with C-terminal amidation (Fig. 1.1). NPY is one of the most abundant neuropeptides in the brain (Gray and Morley, 1986), additionally it is also expressed in the peripheral nervous system and plays an important role in the regulation of many physiological processes, such as food intake, thermogenesis, mood, memory, blood pressure and reproduction (Berglund et al., 2003). At the same time NPY is also extremely well conserved among species (Larhammar, 1996). By contrast, PYY shows greater variability, and PP is the most rapidly evolving member of the NPY peptide family with only 50 % identity within mammals. PYY and PP are hormones that are released in the gastrointestinal tract in response to meals to regulate pancreatic and gastric secretion (Tatemoto, 1982; Hazelwood, 1993). PYY is additionally present in neurons (Ekblad and Sundler, 2002). Each of the NPY receptors shows its individual ligand binding profile for the members of the NPY family of peptides (Table 1.1).

1 10 20 30

hNPY YPSKPDNPGEDAPAEDMARYYSALRHYINLITRQRY-NH2

hPYY YPIKPEAPGEDASPEELNRYYASLRHYLNLVTRQRY-NH2

hPP APLEPVYPGDNATPEQMAQYAADLRRYINMLTRPRY-NH2

Fig. 1.1: Alignment of the human NPY, PYY and PP amino acid sequence; Constant positions in all species for the three peptides are underlined. The seven constant residues within the NPY-family are indicated (boxed).

(17)

General introduction 3 1.2.2 NPY receptors

NPY receptors belong to the rhodopsin like GPCR superfamily and have a common main signaling pathway via Gi/o proteins (Michel et al., 1998). Subtypes in humans comprise the Y1, Y2, Y4, Y5 and y6, with the y6 being non-functional because of a frameshift mutation (single base deletion) in the third intracellular loop leading to a truncated receptor protein after the 6th transmembrane region (Rose et al., 1997). The average sequence homology between the subtypes is relatively low (27-31 %). NPY receptors play a role in a plethora of physiological processes. Some examples of current NPY investigation fields are their implication in depression (Painsipp et al., 2009a; Painsipp et al., 2009b), alcohol dependence (Thorsell, 2007; Wetherill et al., 2008) or pain modulation (Gibbs and Hargreaves, 2008).

Besides the pharmacological aspect as a target the Y1 receptor e.g. is also regarded as a tumor marker in breast cancer as it is overexpressed in malignant tissues (Amlal et al., 2006).

However, the most prominent effect mediated by the NPY multiligand/multireceptor system still remains its involvement in regulating energy homeostasis. For example, Y1 or Y5

receptor selective agonists stimulate feeding (Gerald et al., 1996; Mullins et al., 2001; Henry et al., 2005) and antagonists produce anti-obesity effects. In contrast, Y2 or Y4 receptor selective agonists show inhibition of food intake (Batterham et al., 2002; Asakawa et al., 2003), though these findings were controversially discussed (Tschöp et al., 2004). As obesity and resulting co-morbidities are increasing health problems there has been a special interest in NPY receptor ligands (cf. section 1.2.3). In Table 1.1 an overview of the most important properties of NPY receptors is given (for a survey of Y receptor antagonists see section 1.2.3.)

Table 1.1: Overview of NPY receptors adapted from (Merten and Beck-Sickinger, 2006) Y1R

Expression pattern Signal transduction Physiological functions Ligand binding profile (agonists)

Selective agonists

Cerebral cortex, vascular smooth muscle cells, colon, human adipocytes

Gi/o → cAMP↓; [Ca2+]↑

Analgesia, anxiolysis, circadian rhythm regulation, endocrine regulation, increase feeding, sedative, vasoconstriction NPY ≈ PYY ≈ [Leu31,Pro34]NPY > NPY2-36 ≈ NPY3-36 ≥ PP ≈ NPY13-36

[Phe7,Pro34]NPY; [Leu31,Pro34]NPY/PYY; [Arg6,Pro34]NPY

(18)

Y2R

Expression pattern Signal transduction Physiological functions Ligand binding profile (agonists)

Selective agonists

Nerve fibers, hippocampus, intestine, blood vessels Gi/o → cAMP↓; [Ca2+]↑

Angiogenesis, anxiogenesis, enhanced memory, decreased neurotransmitter secretion, decrease feeding, anticonvulsant PYY > PYY3-36 ≈ NPY3-36 ≈ NPY2-36 ≈ NPY13-36 >>

[Leu31,Pro34]NPY

NPY3-36; NPY13-36; [Ahx5-24]NPY Y4R

Expression pattern Signal transduction Physiological functions Ligand binding profile (agonists)

Selective agonists

Hypothalamus, skeletal muscle, thyroid gland, stomach, small intestine, colon

Gi/o → cAMP↓; [Ca2+]↑

Pancreatic secretion, gall bladder contraction, LH secretion, decrease feeding

PP ≥ GW1229 > PYY ≥ NPY > NPY2-36

PP; GW1229 Y5R

Expression pattern Signal transduction Physiological functions Ligand binding profile (agonists)

Selective agonists

Hypothalamus, cerebral cortex, intestine, ovary, spleen, pancreas, skeletal muscle

Gi/o → cAMP↓; [Ca2+]↑

Circadian rhythm regulation, increase feeding, anticonvulsant, reproduction

NPY ≈ PYY ≈ NPY2-36 ≈ [Leu31,Pro34]NPY > hPP >

[D-Trp32]NPY > NPY13-36 > rPP

[Ala31,Aib32]NPY; [Leu31,Pro34]NPY; [D-Trp34]NPY

(19)

General introduction 5 1.2.3 NPY receptor ligands

Starting from the endogenous ligands, NPY, PYY and PP, structure-affinity/activity relationships have been intensely studied by peptide modifications such as amino acid replacements, truncations, cyclizations or building chimera. Thereby, selective peptidergic agonists (listed in Table 1.1) have been widely accepted as reference compounds for the characterization of NPY receptors (Beck-Sickinger et al., 1992; Cabrele and Beck-Sickinger, 2000; Cabrele et al., 2000). Likewise, essential structures from the endogenous ligands were identified for the binding to each receptor. Thus, the small-molecule antagonist reported for the Y1R, BIBP 3226 (Rudolf et al., 1994) (Fig. 1.2), is considered a mimic of the C-terminus, i.e. Arg35 and Tyr36 in NPY (Sautel et al., 1996). The first non-peptidic antagonist was HE- 90481 (Michel and Motulsky, 1990). Likewise, BIIE 0246 (Doods et al., 1999; Dumont et al., 2000) and CGP 71683A (Criscione et al., 1998) (Fig. 1.3) represented the first selective and potent antagonists for the Y2R and the Y5R, respectively. In the following, an overview of the development of Y receptor ligands of the last five years is given. As reviewed by Sato et al.

(2009b), the major motivation to design new compounds for neuropeptide Y receptors was the treatment of obesity. Due to the lack of oral availability and inability to penetrate across the blood brain barrier NPY receptors could not be easily addressed in vivo with the classical substances. However these have been useful pharmacological tools for the characterization of YRs.

Fig. 1.2 shows a selection of developed novel potent and selective Y1 receptor antagonists, all of which show promising results in the inhibition of feeding and the reduction of body weight in animal models, however only if applied systemically, i.e. intracerebroventricularly, intravenously or intraperitoneally (i.c.v., i.v. or i.p.). In order to advance into clinical trial, there is still a need for substances with suitable pharmacokinetic profiles and physicochemical properties.

Beside the aim to treat obesity several compounds have been developed as pharmacological tools. For example, Keller et al. designed a tritium labeled NG-propionylargininamide ([3H]- UR-MK114) that is a BIBP 3226 derivative (Keller et al., 2008). Moreover, there have been reports on agonists with peptidic structures for PET or SPECT imaging of breast cancer (Guerin et al., 2010; Khan et al., 2010) and a carboxyfluorescein labeled small peptide derivative for tumour diagnostics and therapy (Zwanziger et al., 2009).

(20)

Fig. 1.2: Selection of non-peptidic Y1 antagonist; a(Rudolf et al., 1994), b(Wieland et al., 1998), c(Kanatani et al., 2001), d(Leslie et al., 2007), e(Antal-Zimanyi et al., 2008), f(Kameda et al., 2009)

Several novel compounds different in structure from the peptidomimetic BIIE 0246 have been designed for the Y2 receptor (Fig. 1.3). Here the situation is similar as for the Y1R, namely the development of many potent and selective substances has been successful (for review see (Sato et al., 2009b). These compounds were demonstrated to penetrate into the brain (Brothers et al., 2010) when administered i.p. or subcutaneuosly (s.c.) Apart from the synthesis of small molecules the attempt has been pursued to conjugate peptidic Y2R agonists (based on the C-terminus of PYY) with polyethylene glycol (20 kDa) to achieve a higher stability and to modify the amino acid sequence for higher selectivity. Indeed, the substances showed reduction of food intake and body weight (DeCarr et al., 2007; Lumb et al., 2007;

Ortiz et al., 2007). Obinepitide, a Y2/Y4 dual peptide agonist is reported to be in clinical trial (Sato et al., 2009b).

(21)

General introduction 7

Fig. 1.3: Selection of non-peptidic Y2 antagonists; a(Doods et al., 1999), b(Bonaventure et al., 2004), c(Brothers et al., 2010), d(Shoblock et al., 2010)

For the Y4 receptor only UR-AK49, a weak antagonist, has been reported up to date (Ziemek et al., 2007) (Fig. 1.4). In Table 1.1 GW1229, based on the C-terminus of NPY, is mentioned as a selective and potent Y4R agonist (Parker et al., 1998; Schober et al., 1998), although this compound is also active at the Y1Rasan antagonist. Furthermore, recent studies are focusing on peptidergic agonists as the Y4R is known to mediate satiety via the release of PP in response to meals. For example, Sub[-Tyr-Arg-Leu-Arg-Tyr-NH2]2 - a peptide dimer based on NPY32-36 - has been reported to be a selective and highly potent Y4 agonist, which inhibited food intake in mice after i.p. application (Balasubramaniam et al., 2006). As already mentioned for the Y2, the strategy to address Y2 and Y4 receptors at the same time with selective peptidergic agonists is pursued (Balasubramaniam et al., 2007).

Fig. 1.4: UR-AK49, an acylguanidine with moderate Y4 antagonistic activity; aInhibition of hPP induced luminescence in CHO cells expressing the hY4R, the chimeric G protein Gqi5 and mitochondrially targeted apoaequorin.

NH

NH H2N NH

O HN

N N

N O

O N

O O

N

*

HN

O BIIE 0246

IC50 = 3.3 nMa

N O CH3 N

O

CN N

JNJ-5207787 IC50 = 100 nMb ([125I]PYY)

OEt HN

N S HO

SF-11 Ki = 1.55 nMc

HN O

SO2 HN Cl CH3

CH3 SF-21

Ki = 1.93 nMc

N O N CH3

CH3 N

HN O

N F

JNJ-31020028 IC50 = 9 nMd ([125I]PYY)

(22)

The first non-peptidic Y5 antagonist reported was CGP 71683A (Criscione et al., 1998), whereas MK-0557 (Erondu et al., 2006) was the first substance to be applied in clinical trial.

As MK-0557 was well tolerated and showed small, but statistically significant reduction in body weight, several new compounds were designed (Fig. 1.5), (Ando et al., 2009; Haga et al., 2009; Sato et al., 2009a) with promising pharmacological and pharmacokinetic properties so that proof-of-concept studies in humans are planned. Due to its expression in the limbic system, the Y5R was hypothesized to modulate stress sensitivity. Indeed, Lu AA33810 was shown to exert anxiolytic- and antidepressant-like effects in rats (Walker et al., 2009).

Fig. 1.5: Selection of Y5R antagonists ; a(Criscione et al., 1998), b(Erondu et al., 2006), c(Ando et al., 2009),

d(Sato et al., 2009a), e(Walker et al., 2009)

1.3 Development of novel functional assays for NPY receptors

Principally, for the pharmacological characterization of GPCR ligands, such as the substances presented in the previous section, in addition to binding data, the determination of the quality of action, agonistic potency or antagonistic activity is a must. There are numerous methods to determine potency and efficacy of ligands at the GPCR of interest, each of which has its specific applications, advantages and disadvantages. Therefore, a maximum of information on a given GPCR can only be obtained by combining complementary approaches.

For an overview of existing functional assays for NPY receptors the reader is referred to the introductory sections of chapters 4 to 6. This work aims at the development of new assay types presented in the current section exploiting GPCR signaling via various heterotrimeric G proteins.

(23)

General introduction 9 As already mentioned (section 1.1), one of the prevailing signaling pathways of 7TMRs is

coupling to heterotrimeric G proteins. Upon agonist binding a conformational change of the receptor takes place leading to association with a free inactive G protein consisting of a Gα- subunit and a Gβγ heterodimer, located on the cytosolic side of the membrane. Subsequently, Gα-bound GDP is released and replaced by GTP, which causes the dissociation of the ternary complex into the GPCR, the GTP-bound Gα-subunit and the Gβγ-dimer. The G protein subunits each activate effector proteins until the intrinsic GTPase activity of Gα cleaves the nucleotide into GDP and inorganic phosphate (Pi). Termination of the cycle is completed by the association of GDP-bound Gα with the Gβγ-dimer. The inactive G protein heterotrimer is available for another round of activation (Fig. 1.6).

There are four classes of G proteins with respect to the homology of their Gα sequences: Gs, Gi/o, Gq/11 and G12/13 (Cabrera-Vera et al., 2003; Milligan and Kostenis, 2006). Gs proteins activate adenylyl cyclase (AC), which results in the formation of 3´,5´-adenosine mono- phosphate (cAMP). Members of the Gi/o class inhibit AC, while Gq/11 activate phospholipase Cβ (PLCβ), that catalyzes the formation of inositol-1,4,5-triphosphate (IP3) and 1,2-diacylglycerol (DAG). G12/13 proteins interact with guanine nucleotide exchange factors. The modulation of the mentioned second messengers leads to diverse downstream cellular signals, e.g. cAMP activates protein kinase A (PKA) resulting in the phosphorylation of various substrate proteins. Furthermore, exchange proteins directly activated by cAMP (Epac) as well as cAMP-gated ion channels are regulated. The inhibition of cAMP formation prevents these effects. Elevated IP3 levels result in the release of intracellular calcium (Ca2+)

Fig. 1.6: G protein activation/

deactivation cycle after GPCR stimulation by an agonist; Adapted from (Seifert and Wieland, 2005)

(24)

from the endoplasmic reticulum (ER), while DAG stimulates protein kinase C (PKC), which in turn stimulates various intracellular proteins by phosphorylation. Additionally, activated Gβγ-dimers can also trigger cellular effects, for instance, by interacting with AC, PLCβ or certain ion channels.

Gα-subunits as well as Gβγ-heterodimers are anchored to the cell membrane by lipidation of their N-termini. The interaction of Gα with a GPCR is defined by the amino acid sequence of its C-terminus, which is recognized by the receptor.

A large and diverse family of proteins – the regulators of G protein signaling (RGS) – has its part in the G protein cycle by accelerating the GTPase activity of the Gα-subunit (Ross and Wilkie, 2000; Willars, 2006) by stabilization of the transition state of GTP cleavage. Thus, RGS proteins function as negative regulators of G protein signaling.

1.3.1 FRET based cAMP assay for the human NPY Y1 receptor

In case of the NPY receptors, coupling to Gi/o proteins, the inhibition of cAMP formation has to be measured to determine receptor activation. Therefore, pre-stimulation with e.g.

forskolin, a direct activator of adenylyl cyclase, is required to see an effect. Classical assay formats for YRs are based, for example, on measuring cAMP radioimmunologically or spectroscopically after enzymatic conversion of the second messenger to NADPH. To avoid such hazardous and laborious approaches in monitoring cAMP levels, several sensors have been developed from structures of known interaction partners of the second messenger, which are, e.g., exchange protein, directly activated by cAMP 1 (Epac1) and Epac2 (de Rooij et al., 1998; Bos, 2003). Such proteins contain a catalytic domain regulated by a cAMP binding site.

Upon binding of the second messenger, the conformation of Epac is changed, releasing the catalytic part. Because of this unfolding of the protein (the termini are supposed to move away from each other) the cyan and yellow fluorescent proteins (CFP and YFP) have been fused to the ends and cAMP has been measured by means of fluorescence resonance energy transfer (FRET) (Ponsioen et al., 2004). For FRET to take place, the applied fluorophores have to overlap in their emission and absorption spectra, as shown for CFP and YFP as an example in Fig. 1.7 A. Moreover, they have to be in close proximity to each other, i.e.

1-10 nm, which means that in case of CFP-Epac-YFP, the FRET efficiency decreases upon cAMP formation. A further development, devoid of the catalytic domain of Epac, is the Epac cAMP sensor (Epac-camps) (Nikolaev et al., 2004; Nikolaev et al., 2005), which contains a cAMP binding site flanked by CFP and YFP (Fig. 1.7 B). This protein was used in an

(25)

General introduction 11 approach to establish a FRET based assay in SK-N-MC cells endogenously expressing the

hY1R (chapter 3).

1.3.2 Fluorescence and luminescence based calcium assays for the human NPY Y5 receptor

Conklin et al (Conklin et al., 1993) have shown that the G protein specificity of a GPCR depends on its C-terminal amino acid sequence. Subsequently, several G protein chimera were constructed, e.g., consisting of Gαq with the last amino acids replaced by those of Gαi2

(Conklin et al., 1996), thus being able to re-direct receptor signaling to the PLC pathway.

Such chimeric G proteins have already been successfully applied in high throughput screening (HTS) at Gi/o coupling receptors (Coward et al., 1999; Dautzenberg, 2005; Dautzenberg et al., 2005). Ca2+ chelating dyes such as fluo-4 or the ratiometric fura-2 are used to measure the intracellular Ca2+ mobilization in flow cytometric or spectrofluorimetric assays as already established, e.g., for the hY2 and the hY4 receptors, stably co-expressed with Gαqi5 in CHO cells (Ziemek et al., 2006; Ziemek et al., 2007). Because such fluorescence based assays suffer from dye leakage from the cells, which results in decreased signal-to-noise ratios during experiments, another approach based on luminescence with the photoprotein aequorin targeted to the mitochondria (see below) is favorable (Ziemek et al., 2006; Ziemek et al., 2007). Aequorin is a luciferase, which is reconstituted as a holoprotein with its chromophore cofactor coelenterazine (a luciferin). Upon binding of Ca2+, coelenterazine is oxidized to coelenteramide, and CO2, and luminescence occurs (λmax = 470 nm) (Shimomura and Johnson, 1978; Jones et al., 1999). The advantages of aequorin as an intracellular calcium indicator are its high sensitivity, low background, as well as absence of toxicity and its large linear dynamic range (Dupriez et al., 2002). For functional GPCR assays cytoplasmically expressed and mitochondrially targeted aequorin (cytAEQ and mtAEQ) have been applied,

A B

Fig. 1.7: Absorbance and emission spectra of CFP and YFP (A) and the principle of Epac-camps (B)

(26)

both giving the same functional data (Stables et al., 1997; Stables et al., 2000), whereas substantially stronger luminescence was obtained with mtAEQ.

1.3.3 Steady-state GTPase assay for the human NPY Y2 and Y4 receptors

The steady-state GTPase assay aims at directly measuring receptor activation, which subsequently results in the activation of the G protein cycle and the cleavage of [γ-32P]GTP.

This is favorable for GPCRs coupling to Gi/o proteins, such as the NPY receptors, as no pre- stimulation is necessary.

For such assays membranes containing the respective GPCR, appropriate G proteins are required and in some cases the coexpression of RGS proteins turned out to be useful. To generate such membranes, the baculovirus/Sf9 cell expression system is used, which is well established for many GPCRs (Massotte, 2003; Aloia et al., 2009). The Sf9 cell line is derived from Spodoptera frugiperda ovarian tissue and can be infected by the highly species-specific baculovirus Autographa californica nuclear polyhedrosis virus (AcNPV), which was used for expression purposes. Recombinant baculovirus containing receptor cDNA had to be generated. Because the genome of AcNPV is too large to easily insert foreign genes, a transfer vector (pVL1392) is used. The foreign gene is inserted into the vector in such a way that its expression is controlled by the strong late phase polyhedrin promoter. Polyhedrin, the matrix protein in which virus particles are embedded, can be replaced by the cDNA of interest. Upon co-transfection into Sf9 cells (Fig.1.8), the transfer vector and the linearized baculovirus DNA undergo homologous recombination, yielding viable recombinant baculovirus.

Fig. 1.8: Generation of recombinant hYxR-ba- culoviruses and protein expression

(27)

General introduction 13 By the infection of Sf9 cells with such viruses high GPCR or G protein expression levels can

be achieved (Seifert et al., 1998; Massotte, 2003; Schneider et al., 2009). Co-infection with up to four baculoviruses, thereby combining the receptor of interest with diverse Gα-, Gβγ- subunits and RGS proteins, is tolerated by the cells (Kleemann et al., 2008). Thus, for example G protein selectivity of the GPCR of interest can be studied.

From functional experiments and immunoblot analyses it can be concluded that Sf9 cells do not express any constitutively active GPCRs, i.e. receptors, that isomerize to the activated form in the absence of agonists, and that signaling of mammalian GPCRs via endogenous G protein is limited (Quehenberger et al., 1992; Wenzel-Seifert et al., 1998; Brys et al., 2000;

Seifert et al., 2003). This was considered as an excellent starting point for the establishment of functional assays with high signal-to-noise ratios for the molecular pharmacological investigation of NPY receptor ligands and for studying constitutive receptor activity.

1.4 References

Aloia AL, Glatz RV, McMurchie EJ and Leifert WR (2009) GPCR expression using baculovirus-infected Sf9 cells. Methods Mol Biol 552:115-129.

Amlal H, Faroqui S, Balasubramaniam A and Sheriff S (2006) Estrogen up-regulates neuropeptide Y Y1 receptor expression in a human breast cancer cell line. Cancer Res 66:3706-3714.

Ando M, Sato N, Nagase T, Nagai K, Ishikawa S, Takahashi H, Ohtake N, Ito J, Hirayama M, Mitobe Y, Iwaasa H, Gomori A, Matsushita H, Tadano K, Fujino N, Tanaka S, Ohe T, Ishihara A, Kanatani A and Fukami T (2009) Discovery of pyridone-containing imidazolines as potent and selective inhibitors of neuropeptide Y Y5 receptor. Bioorg Med Chem 17:6106-6122.

Antal-Zimanyi I, Bruce MA, Leboulluec KL, Iben LG, Mattson GK, McGovern RT, Hogan JB, Leahy CL, Flowers SC, Stanley JA, Ortiz AA and Poindexter GS (2008) Pharmacological characterization and appetite suppressive properties of BMS-193885, a novel and selective neuropeptide Y1 receptor antagonist. Eur J Pharmacol 590:224- 232.

Asakawa A, Inui A, Yuzuriha H, Ueno N, Katsuura G, Fujimiya M, Fujino MA, Niijima A, Meguid MM and Kasuga M (2003) Characterization of the effects of pancreatic polypeptide in the regulation of energy balance. Gastroenterology 124:1325-1336.

Balasubramaniam A, Joshi R, Su C, Friend LA and James JH (2007) Neuropeptide Y (NPY) Y2 receptor-selective agonist inhibits food intake and promotes fat metabolism in mice: combined anorectic effects of Y2 and Y4 receptor-selective agonists. Peptides 28:235-240.

Balasubramaniam A, Mullins DE, Lin S, Zhai W, Tao Z, Dhawan VC, Guzzi M, Knittel JJ, Slack K, Herzog H and Parker EM (2006) Neuropeptide Y (NPY) Y4 receptor selective agonists based on NPY(32-36): development of an anorectic Y4 receptor selective agonist with picomolar affinity. J Med Chem 49:2661-2665.

(28)

Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, Wren AM, Brynes AE, Low MJ, Ghatei MA, Cone RD and Bloom SR (2002) Gut hormone PYY(3-36) physiologically inhibits food intake. Nature 418:650-654.

Beck-Sickinger AG, Grouzmann E, Hoffmann E, Gaida W, van Meir EG, Waeber B and Jung G (1992) A novel cyclic analog of neuropeptide Y specific for the Y2 receptor. Eur J Biochem 206:957-964.

Berglund MM, Hipskind PA and Gehlert DR (2003) Recent developments in our understanding of the physiological role of PP-fold peptide receptor subtypes. Exp Biol Med (Maywood) 228:217-244.

Bockaert J and Pin JP (1999) Molecular tinkering of G protein-coupled receptors: an evolutionary success. EMBO J 18:1723-1729.

Bonaventure P, Nepomuceno D, Mazur C, Lord B, Rudolph DA, Jablonowski JA, Carruthers NI and Lovenberg TW (2004) Characterization of N-(1-Acetyl-2,3-dihydro-1H-indol- 6-yl)-3-(3-cyano-phenyl)-N-[1-(2-cyclopen tyl-ethyl)-piperidin-4yl]acrylamide (JNJ- 5207787), a small molecule antagonist of the neuropeptide Y Y2 receptor. J Pharmacol Exp Ther 308:1130-1137.

Bos JL (2003) Epac: a new cAMP target and new avenues in cAMP research. Nat Rev Mol Cell Biol 4:733-738.

Brothers SP, Saldanha SA, Spicer TP, Cameron M, Mercer BA, Chase P, McDonald P, Wahlestedt C and Hodder PS (2010) Selective and brain penetrant neuropeptide Y Y2

receptor antagonists discovered by whole-cell high-throughput screening. Mol Pharmacol 77:46-57.

Brys R, Josson K, Castelli MP, Jurzak M, Lijnen P, Gommeren W and Leysen JE (2000) Reconstitution of the human 5-HT1D receptor-G-protein coupling: evidence for constitutive activity and multiple receptor conformations. Mol Pharmacol 57:1132- 1141.

Cabrele C and Beck-Sickinger AG (2000) Molecular characterization of the ligand-receptor interaction of the neuropeptide Y family. J Pept Sci 6:97-122.

Cabrele C, Langer M, Bader R, Wieland HA, Doods HN, Zerbe O and Beck-Sickinger AG (2000) The first selective agonist for the neuropeptide Y Y5 receptor increases food intake in rats. J Biol Chem 275:36043-36048.

Cabrera-Vera TM, Vanhauwe J, Thomas TO, Medkova M, Preininger A, Mazzoni MR and Hamm HE (2003) Insights into G Protein Structure, Function, and Regulation. Endocr Rev 24:765-781.

Conklin BR, Farfel Z, Lustig KD, Julius D and Bourne HR (1993) Substitution of three amino acids switches receptor specificity of Gqα to that of Giα. Nature 363:274-276.

Conklin BR, Herzmark P, Ishida S, Voyno-Yasenetskaya TA, Sun Y, Farfel Z and Bourne HR (1996) Carboxyl-terminal mutations of Gqα and Gsα that alter the fidelity of receptor activation. Mol Pharmacol 50:885-890.

Coward P, Chan SD, Wada HG, Humphries GM and Conklin BR (1999) Chimeric G proteins allow a high-throughput signaling assay of Gi-coupled receptors. Anal Biochem 270:242-248.

Criscione L, Rigollier P, Batzl-Hartmann C, Rueger H, Stricker-Krongrad A, Wyss P, Brunner L, Whitebread S, Yamaguchi Y, Gerald C, Heurich RO, Walker MW, Chiesi M, Schilling W, Hofbauer KG and Levens N (1998) Food intake in free-feeding and energy-deprived lean rats is mediated by the neuropeptide Y5 receptor. J Clin Invest 102:2136-2145.

Dautzenberg FM (2005) Stimulation of neuropeptide Y-mediated calcium responses in human SMS-KAN neuroblastoma cells endogenously expressing Y2 receptors by co- expression of chimeric G proteins. Biochem Pharmacol 69:1493-1499.

(29)

General introduction 15 Dautzenberg FM, Higelin J, Pflieger P, Neidhart W and Guba W (2005) Establishment of

robust functional assays for the characterization of neuropeptide Y (NPY) receptors:

identification of 3-(5-benzoyl-thiazol-2-ylamino)-benzonitrile as selective NPY type 5 receptor antagonist. Neuropharmacology 48:1043-1055.

de Rooij J, Zwartkruis FJ, Verheijen MH, Cool RH, Nijman SM, Wittinghofer A and Bos JL (1998) Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP. Nature 396:474-477.

DeCarr LB, Buckholz TM, Milardo LF, Mays MR, Ortiz A and Lumb KJ (2007) A long- acting selective neuropeptide Y2 receptor PEGylated peptide agonist reduces food intake in mice. Bioorg Med Chem Lett 17:1916-1919.

Doods H, Gaida W, Wieland HA, Dollinger H, Schnorrenberg G, Esser F, Engel W, Eberlein W and Rudolf K (1999) BIIE0246: a selective and high affinity neuropeptide Y Y2

receptor antagonist. Eur J Pharmacol 384:R3-5.

Dumont Y, Cadieux A, Doods H, Pheng LH, Abounader R, Hamel E, Jacques D, Regoli D and Quirion R (2000) BIIE0246, a potent and highly selective non-peptide neuropeptide Y Y2 receptor antagonist. Br J Pharmacol 129:1075-1088.

Dupriez VJ, Maes K, Le Poul E, Burgeon E and Detheux M (2002) Aequorin-based functional assays for G-protein-coupled receptors, ion channels, and tyrosine kinase receptors. Receptors Channels 8:319-330.

Ekblad E and Sundler F (2002) Distribution of pancreatic polypeptide and peptide YY.

Peptides 23:251-261.

Erondu N, Gantz I, Musser B, Suryawanshi S, Mallick M, Addy C, Cote J, Bray G, Fujioka K, Bays H, Hollander P, Sanabria-Bohorquez SM, Eng W, Langstrom B, Hargreaves RJ, Burns HD, Kanatani A, Fukami T, MacNeil DJ, Gottesdiener KM, Amatruda JM, Kaufman KD and Heymsfield SB (2006) Neuropeptide Y5 receptor antagonism does not induce clinically meaningful weight loss in overweight and obese adults. Cell Metab 4:275-282.

Fredriksson R, Lagerström MC, Lundin LG and Schiöth HB (2003) The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol Pharmacol 63:1256-1272.

Gerald C, Walker MW, Criscione L, Gustafson EL, Batzl-Hartmann C, Smith KE, Vaysse P, Durkin MM, Laz TM, Linemeyer DL, Schaffhauser AO, Whitebread S, Hofbauer KG, Taber RI, Branchek TA and Weinshank RL (1996) A receptor subtype involved in neuropeptide-Y-induced food intake. Nature 382:168-171.

Gibbs JL and Hargreaves KM (2008) Neuropeptide Y Y1 receptor effects on pulpal nociceptors. J Dent Res 87:948-952.

Gray TS and Morley JE (1986) Neuropeptide Y: anatomical distribution and possible function in mammalian nervous system. Life Sci 38:389-401.

Guerin B, Dumulon-Perreault V, Tremblay MC, Ait-Mohand S, Fournier P, Dubuc C, Authier S and Benard F (2010) [Lys(DOTA)4]BVD15, a novel and potent neuropeptide Y analog designed for Y1 receptor-targeted breast tumor imaging. Bioorg Med Chem Lett 20:950-953.

Haga Y, Sakamoto T, Shibata T, Nonoshita K, Ishikawa M, Suga T, Takahashi H, Takahashi T, Ando M, Murai T, Gomori A, Oda Z, Kitazawa H, Mitobe Y, Kanesaka M, Ohe T, Iwaasa H, Ishii Y, Ishihara A, Kanatani A and Fukami T (2009) Discovery of trans-N- [1-(2-fluorophenyl)-3-pyrazolyl]-3-oxospiro[6-azaisobenzofuran-1(3H),1'-

cyclohexane]-4'-carboxamide, a potent and orally active neuropeptide Y Y5 receptor antagonist. Bioorg Med Chem 17:6971-6982.

Hazelwood RL (1993) The pancreatic polypeptide (PP-fold) family: gastrointestinal, vascular, and feeding behavioral implications. Proc Soc Exp Biol Med 202:44-63.

(30)

Henry M, Ghibaudi L, Gao J and Hwa JJ (2005) Energy metabolic profile of mice after chronic activation of central NPY Y1, Y2, or Y5 receptors. Obes Res 13:36-47.

Jaakola VP, Griffith MT, Hanson MA, Cherezov V, Chien EY, Lane JR, Ijzerman AP and Stevens RC (2008) The 2.6 angstrom crystal structure of a human A2A adenosine receptor bound to an antagonist. Science 322:1211-1217.

Jacoby E, Bouhelal R, Gerspacher M and Seuwen K (2006) The 7 TM G-protein-coupled receptor target family. ChemMedChem 1:761-782.

Jones K, Hibbert F and Keenan M (1999) Glowing jellyfish, luminescence and a molecule called coelenterazine. Trends Biotechnol 17:477-481.

Kameda M, Kobayashi K, Ito H, Miyazoe H, Tsujino T, Nakama C, Kawamoto H, Ando M, Ito S, Suzuki T, Kanno T, Tanaka T, Tahara Y, Tani T, Tanaka S, Tokita S and Sato N (2009) Optimization of a series of 2,4-diaminopyridines as neuropeptide Y Y1 receptor antagonists with reduced hERG activity. Bioorg Med Chem Lett 19:4325-4329.

Kanatani A, Hata M, Mashiko S, Ishihara A, Okamoto O, Haga Y, Ohe T, Kanno T, Murai N, Ishii Y, Fukuroda T, Fukami T and Ihara M (2001) A typical Y1 receptor regulates feeding behaviors: effects of a potent and selective Y1 antagonist, J-115814. Mol Pharmacol 59:501-505.

Keller M, Pop N, Hutzler C, Beck-Sickinger AG, Bernhardt G and Buschauer A (2008) Guanidine-acylguanidine bioisosteric approach in the design of radioligands: synthesis of a tritium-labeled NG-propionylargininamide ([3H]-UR-MK114) as a highly potent and selective neuropeptide Y Y1 receptor antagonist. J Med Chem 51:8168-8172.

Khan IU, Zwanziger D, Bohme I, Javed M, Naseer H, Hyder SW and Beck-Sickinger AG (2010) Breast-cancer diagnosis by neuropeptide Y analogues: from synthesis to clinical application. Angew Chem Int Ed Engl 49:1155-1158.

Kleemann P, Papa D, Vigil-Cruz S and Seifert R (2008) Functional reconstitution of the human chemokine receptor CXCR4 with Gi/Go-proteins in Sf9 insect cells. Naunyn Schmiedebergs Arch Pharmacol 378:261-274.

Lagerström MC and Schiöth HB (2008) Structural diversity of G protein-coupled receptors and significance for drug discovery. Nat Rev Drug Discov 7:339-357.

Larhammar D (1996) Evolution of neuropeptide Y, peptide YY and pancreatic polypeptide.

Regul Pept 62:1-11.

Leslie CP, Di Fabio R, Bonetti F, Borriello M, Braggio S, Dal Forno G, Donati D, Falchi A, Ghirlanda D, Giovannini R, Pavone F, Pecunioso A, Pentassuglia G, Pizzi DA, Rumboldt G and Stasi L (2007) Novel carbazole derivatives as NPY Y1 antagonists.

Bioorg Med Chem Lett 17:1043-1046.

Lumb KJ, DeCarr LB, Milardo LF, Mays MR, Buckholz TM, Fisk SE, Pellegrino CM, Ortiz AA and Mahle CD (2007) Novel selective neuropeptide Y2 receptor PEGylated peptide agonists reduce food intake and body weight in mice. J Med Chem 50:2264- 2268.

Massotte D (2003) G protein-coupled receptor overexpression with the baculovirus-insect cell system: a tool for structural and functional studies. Biochimica et Biophysica Acta 1610:77-89.

Merten N and Beck-Sickinger AG (2006) Molecular ligand-receptor interaction of the NPY/PP peptide family. EXS:35-62.

Michel MC, Beck-Sickinger A, Cox H, Doods HN, Herzog H, Larhammar D, Quirion R, Schwartz T and Westfall T (1998) XVI. International Union of Pharmacology recommendations for the nomenclature of neuropeptide Y, peptide YY, and pancreatic polypeptide receptors. Pharmacol Rev 50:143-150.

Michel MC and Motulsky HJ (1990) HE 90481: A competitive nonpeptidergic antagonist at neuropeptide Y receptors. Ann N Y Acad Sci 611:392-394.

(31)

General introduction 17 Milligan G and Kostenis E (2006) Heterotrimeric G-proteins: a short history. Br J Pharmacol

147 Suppl 1:S46-55.

Mullins D, Kirby D, Hwa J, Guzzi M, Rivier J and Parker E (2001) Identification of potent and selective neuropeptide Y Y1 receptor agonists with orexigenic activity in vivo.

Mol Pharmacol 60:534-540.

Nikolaev VO, Bunemann M, Hein L, Hannawacker A and Lohse MJ (2004) Novel single chain cAMP sensors for receptor-induced signal propagation. J Biol Chem 279:37215- 37218.

Nikolaev VO, Gambaryan S, Engelhardt S, Walter U and Lohse MJ (2005) Real-time monitoring of the PDE2 activity of live cells: hormone-stimulated cAMP hydrolysis is faster than hormone-stimulated cAMP synthesis. J Biol Chem 280:1716-1719.

Ortiz AA, Milardo LF, DeCarr LB, Buckholz TM, Mays MR, Claus TH, Livingston JN, Mahle CD and Lumb KJ (2007) A novel long-acting selective neuropeptide Y2

receptor polyethylene glycol-conjugated peptide agonist reduces food intake and body weight and improves glucose metabolism in rodents. J Pharmacol Exp Ther 323:692- 700.

Painsipp E, Herzog H and Holzer P (2009a) Evidence from knockout mice that neuropeptide- Y Y2 and Y4 receptor signaling prevents long-term depression-like behaviour caused by immune challenge. J Psychopharmacol.

Painsipp E, Sperk G, Herzog H and Holzer P (2009b) Delayed stress-induced differences in locomotor and depression-related behaviour in female neuropeptide-Y Y1 receptor knockout mice. J Psychopharmacol.

Palczewski K, Kumasaka T, Hori T, Behnke CA, Motoshima H, Fox BA, Le Trong I, Teller DC, Okada T, Stenkamp RE, Yamamoto M and Miyano M (2000) Crystal structure of rhodopsin: A G protein-coupled receptor. Science 289:739-745.

Parker EM, Babij CK, Balasubramaniam A, Burrier RE, Guzzi M, Hamud F, Mukhopadhyay G, Rudinski MS, Tao Z, Tice M, Xia L, Mullins DE and Salisbury BG (1998) GR231118 (1229U91) and other analogues of the C-terminus of neuropeptide Y are potent neuropeptide Y Y1 receptor antagonists and neuropeptide Y Y4 receptor agonists. Eur J Pharmacol 349:97-105.

Ponsioen B, Zhao J, Riedl J, Zwartkruis F, van der Krogt G, Zaccolo M, Moolenaar WH, Bos JL and Jalink K (2004) Detecting cAMP-induced Epac activation by fluorescence resonance energy transfer: Epac as a novel cAMP indicator. EMBO Rep 5:1176-1180.

Quehenberger O, Prossnitz ER, Cochrane CG and Ye RD (1992) Absence of Gi proteins in the Sf9 insect cell. Characterization of the uncoupled recombinant N-formyl peptide receptor. J Biol Chem 267:19757-19760.

Rajagopal S, Rajagopal K and Lefkowitz RJ (2010) Teaching old receptors new tricks:

biasing seven-transmembrane receptors. Nat Rev Drug Discov 9:373-386.

Rasmussen SG, Choi HJ, Rosenbaum DM, Kobilka TS, Thian FS, Edwards PC, Burghammer M, Ratnala VR, Sanishvili R, Fischetti RF, Schertler GF, Weis WI and Kobilka BK (2007) Crystal structure of the human β2 adrenergic G-protein-coupled receptor.

Nature 450:383-387.

Rose PM, Lynch JS, Frazier ST, Fisher SM, Chung W, Battaglino P, Fathi Z, Leibel R and Fernandes P (1997) Molecular genetic analysis of a human neuropeptide Y receptor.

The human homolog of the murine "Y5" receptor may be a pseudogene. J Biol Chem 272:3622-3627.

Rosenbaum DM, Cherezov V, Hanson MA, Rasmussen SG, Thian FS, Kobilka TS, Choi HJ, Yao XJ, Weis WI, Stevens RC and Kobilka BK (2007) GPCR engineering yields high-resolution structural insights into β2-adrenergic receptor function. Science 318:1266-1273.

(32)

Ross EM and Wilkie TM (2000) GTPase-activating proteins for heterotrimeric G proteins:

regulators of G protein signaling (RGS) and RGS-like proteins. Annu Rev Biochem 69:795-827.

Rudolf K, Eberlein W, Engel W, Wieland HA, Willim KD, Entzeroth M, Wienen W, Beck- Sickinger AG and Doods HN (1994) The first highly potent and selective non-peptide neuropeptide Y Y1 receptor antagonist: BIBP3226. Eur J Pharmacol 271:R11-13.

Sato N, Ando M, Ishikawa S, Jitsuoka M, Nagai K, Takahashi H, Sakuraba A, Tsuge H, Kitazawa H, Iwaasa H, Mashiko S, Gomori A, Moriya R, Fujino N, Ohe T, Ishihara A, Kanatani A and Fukami T (2009a) Discovery of tetrasubstituted imidazolines as potent and selective neuropeptide Y Y5 receptor antagonists: reduced human ether-a- go-go related gene potassium channel binding affinity and potent antiobesity effect. J Med Chem 52:3385-3396.

Sato N, Ogino Y, Mashiko S and Ando M (2009b) Modulation of neuropeptide Y receptors for the treatment of obesity. Expert Opin Ther Pat 19:1401-1415.

Sautel M, Rudolf K, Wittneben H, Herzog H, Martinez R, Munoz M, Eberlein W, Engel W, Walker P and Beck-Sickinger AG (1996) Neuropeptide Y and the nonpeptide antagonist BIBP 3226 share an overlapping binding site at the human Y1 receptor. Mol Pharmacol 50:285-292.

Schneider EH, Schnell D, Papa D and Seifert R (2009) High constitutive activity and a G- protein-independent high-affinity state of the human histamine H4-receptor.

Biochemistry 48:1424-1438.

Schober DA, Van Abbema AM, Smiley DL, Bruns RF and Gehlert DR (1998) The neuropeptide Y Y1 antagonist, 1229U91, a potent agonist for the human pancreatic polypeptide-preferring (NPY Y4) receptor. Peptides 19:537-542.

Seifert R, Lee TW, Lam VT and Kobilka BK (1998) Reconstitution of β2-adrenoceptor-GTP- binding-protein interaction in Sf9 cells-high coupling efficiency in a β2-adrenoceptor- Gsα fusion protein. Eur J Biochem 255:369-382.

Seifert R, Wenzel-Seifert K, Burckstummer T, Pertz HH, Schunack W, Dove S, Buschauer A and Elz S (2003) Multiple differences in agonist and antagonist pharmacology between human and guinea pig histamine H1-receptor. Journal of Pharmacology and Experimental Therapeutics 305:1104-1115.

Seifert R and Wieland T (2005) G protein-coupled receptors as drug targets: analysis of activation and constitutive activity. Wiley-VCH, Weinheim.

Shimomura O and Johnson FH (1978) Peroxidized coelenterazine, the active group in the photoprotein aequorin. Proc Natl Acad Sci U S A 75:2611-2615.

Shoblock JR, Welty N, Nepomuceno D, Lord B, Aluisio L, Fraser I, Motley ST, Sutton SW, Morton K, Galici R, Atack JR, Dvorak L, Swanson DM, Carruthers NI, Dvorak C, Lovenberg TW and Bonaventure P (2010) In vitro and in vivo characterization of JNJ- 31020028 (N-(4-{4-[2-(diethylamino)-2-oxo-1-phenylethyl]piperazin-1-yl}-3- fluorophe nyl)-2-pyridin-3-ylbenzamide), a selective brain penetrant small molecule antagonist of the neuropeptide Y Y2 receptor. Psychopharmacology (Berl) 208:265- 277.

Stables J, Green A, Marshall F, Fraser N, Knight E, Sautel M, Milligan G, Lee M and Rees S (1997) A bioluminescent assay for agonist activity at potentially any G-protein- coupled receptor. Anal Biochem 252:115-126.

Stables J, Mattheakis LC, Chang R and Rees S (2000) Recombinant aequorin as reporter of changes in intracellular calcium in mammalian cells. Methods Enzymol 327:456-471.

Tatemoto K (1982) Isolation and characterization of peptide YY (PYY), a candidate gut hormone that inhibits pancreatic exocrine secretion. Proc Natl Acad Sci U S A 79:2514-2518.

Referenzen

ÄHNLICHE DOKUMENTE

This could indicate potential function of the Kelch13 protein in cell cycle proliferation, which would be in line with the observed phenotype of parasite arresting in transition

Abbreviations: WNDP, Wilson disease protein; MNKP, Menkes ATPase; MBS, metal binding site; D1–5WNDP, six-histidine tagged Wilson ATPase only containing metal binding domain 6;..

Although altered expression of CPEBs in gliomas unequivocally indicates the importance of translational regulation in the brain tumors, the exact impact of CPEB dysregulation on

The study of membrane proteins in terms of their structure (e.g. protein crystallization) and their detailed functionality requires the isolation and re-insertion into a

The peroxins can be divided into three groups according to their role in peroxisomal biogenesis: (a) peroxins involved in the import of peroxisomal matrix proteins (1.2.1),

In order to characterize the structural organization of the nuclear export complex composed of CRM1, SPN1 and RanGTP the complex was crystallized (Figure 8-4) and its structure

 Upon  activation  in  the  cytoplasm,  activated  ER  dimers  are  able  to   translocate  into  the  nucleus,  recognize  and  bind  to  estrogen  responsive

Furthermore, structural information on hexameric spastin was obtained through modeling of spastin AAA domain and this modeled structure was used as a framework to classify the